Weekly Digests
‹ Back to April

Release the B7.1! Anti-PD-L1 breaks PD-L1’s grip

April 1, 2020

Blocking the PD-1/PD-L1 axis using anti-PD-L1 antibodies is a commonly employed immunotherapeutic strategy to prevent PD-1-mediated inhibition of T cells by tumor cells. Suspecting that the mechanism of action of PD-L1 blockade might be more complex, however, Mayoux et al. investigated the role of PD-L1-expressing dendritic cells (DCs) in PD-L1 blockade. The results were recently published in Science Translational Medicine.

When a DC primes a T cell, the generally understood mechanism is that B7.1 on dendritic cells engages with the positive costimulator CD28 on T cells, encouraging T cell activation. If the negative costimulator PD-1 is also engaged on T cells, then T cell activation could be inhibited. Mayoux et al. investigated the expression of PD-L1, PD-1, and the CD28 stimulatory ligand B7.1 (aka CD80) on various DC subsets from patients with lung cancer and found that both tumor-associated and peripheral DCs expressed PD-L1. They also observed that tumor-associated but not peripheral cross-presenting DCs (CD141+ and CD1c+) expressed B7.1. Focusing on a double-positive (PD-L1+ B7.1+) population in cross-presenting DCs from fresh renal cell carcinoma samples, and cognizant that PD-L1 and B7.1 have been known to interact in cis, the researchers used an antibody binding assay to quantify surface expression and observed that PD-L1 was 20-fold more abundant than B7.1 on both CD141+ and CD1c+ conventional dendritic cells.

Based on this evidence, Mayoux et al. hypothesized that the abundant PD-L1 could sequester the less abundant B7.1 in cis on DCs, leaving excess PD-L1 available for binding. To test this, they used monocyte-derived DCs from healthy donors in an in vitro system to develop LPS-matured DCs and observed expression patterns of PD-L1 and B7.1 that were similar to those observed in tumors. In a confocal imaging analysis, when the DCs were observed forming synapses with T cells, PD-1 and CD28 colocalized more often than PD-1 and PD-L1, consistent with recent findings that PD-1 signaling may be involved in the dephosphorylation of CD28 at the immunological synapse. Interestingly, B7.1 was barely observed colocalizing with CD28 at all, instead interacting to a much higher degree with PD-L1 in cis, likely due to its higher binding affinity.

To test whether blocking PD-L1 might release B7.1 from sequestration and make it available for interaction with CD28, the researchers modeled the physical interactions between B7.1/PD-L1 and B7.1/CD28. First, they showed that anti-PD-L1 binds to PD-L1-expressing cells with a binding affinity 700-fold higher than the binding affinity between PD-L1 and B7.1, suggesting that the antibody would readily disrupt PD-L1/B7-1 binding. Next, using cells engineered to mimic the expression of B7.1 and PD-L1 on DCs, and CD28-Fc to mimic T cells, the researchers observed an increase in B7.1/CD28 interactions when anti-PD-L1 was present. These results were confirmed in a primary cell-cell system, which also showed that pre-incubating mature DCs with anti-PD-L1 enabled stronger interactions between B7.1 on DCs and CD28 on T cells. Together this evidence indicated that anti-PD-L1 can disrupt the interatctions between PD-L1 and B7.1 in cis, freeing up B7.1, enhancing B7.1 binding to CD28, and activating T cells.

To measure the functional consequences of anti-PD-L1 disrupting the binding between PD-L1 and B7.1 in cis on DCs, Mayoux et al. evaluated CD28 downstream signaling in Jurkat reporter cells. To better isolate the specific contribution of blocking the interaction between PD-L1 and B7.1 in cis, Jurkat cells were first incubated with anti-PD-1 to block PD-1/PD-L1 binding in trans, which increased CD28 signaling by 32%. The cells were then incubated with anti-PD-L1, which further enhanced CD28 signaling by 20%, for an overall increase of 52% in CD28 binding. When the researchers repeated the experiment using a PD-L1-targeting antibody that does not substantially block PD-L1/B7.1 interactions, the increase in CD28 signal transduction was significantly lower.

Next, Mayoux et al. studied the consequences of pretreating activated human DCs with PD-L1 blockade and found that it directly enhanced their ability to stimulate the proliferation of allogeneic T cells. To study whether this effect could generate de novo immune responses, mouse splenic CD11c+ DCs were cocultured with DQ-OVA (cross-presented as the SIINFEKL peptide) and treated with anti-PD-L1. In coculture with naive T cells from OT-I mice, the DCs pretreated with anti-PD-L1 induced more T cell proliferation; increased T-bet and Eomes in T cells, suggesting enhanced T cell effector potential; and enhanced production of granzyme B in T cells in response to target cells. Similar results observed using SIINFEKL peptide-pulsed DCs pretreated with anti-PD-L1.

To test the extent to which tumor-associated DCs contribute to responses to PD-L1 blockade in a clinical setting, Mayoux et al. established a DC gene signature based on RNAseq data and quantification of expression of key genes that defined DC subsets. In patients with renal cell carcinoma treated with atezolizumab (anti-PD-L1), patients with a high pretreatment DC signature showed better overall survival. In patients with non-small cell lung cancer treated with either anti-PD-L1 or docetaxel, a high DC signature correlated with increased overall survival only in patients in the anti-PD-L1 treatment group. Further, the survival benefit was only observed when PD-L1 was expressed on immune cells, refining the correlation between a high DC signature and increased overall survival, and suggesting that these factors could be considered when making decisions regarding treatment options.

Overall, Mayoux et al. show that the activation of T cells by antigen-presenting cells involves a complex network of interactions between PD-1, PD-L1, B7.1, CD28, and possibly CTLA-4 (not yet investigated). They also show that intratumoral DCs expressing PD-L1 and B7.1 likely play a significant role in the outcome of PD-L1 blockade. In addition to blocking interactions between PD-L1 and PD-1, anti-PD-L1 antibodies also prevent interactions between PD-L1 and B7.1 in cis on dendritic cells, freeing up B7.1 to interact more readily with CD28 on T cells and prime T cell activation and antitumor responses – a mechanism not available to anti-PD-1 antibodies. Importantly, the presence of a DC signature could help guide treatment decisions in the clinical setting.

by Lauren Hitchings

References:

Mayoux M., Roller A., Pulko V., Sammicheli S., Chen S., Sum E., Jost C., Fransen M.F., Buser R.B., Kowanetz M., Rommel K., Matos I., Colombetti S., Belousov A., Karanikas V., Ossendorp F., Hegde P.S., Chen D.S., Umana P., Perro M., Klein C., Xu W. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy. Sci Transl Med. 2020 Mar 11.

In the Spotlight...

Long-distance modulation of bystander tumor cells by CD8+ T-cell-secreted IFN-γ

Hoekstra and Bornes et al. created an IFNγ-sensing reporter (ISG) system and used intravital imaging to probe the space/time impact of IFNγ secreted from tumor-infiltrating T cells on ‘antigen (Ag)-loss’ tumor variants. Ag-specific CD8+ T cells were infused into immune deficient mice bearing tumors of intermingled Ag+GFP+ and Ag- ISG+ tumor cells. Even if Ag+ cells were only a small part of the tumor mass, IFNγ signaled IFNγR+Ag-ISG+ cells both near and, with time, distal (over 800 µm; 30-40 cell layers) from antigen encounter sites. IFNγ boosted PD-L1 on Ag-ISG+ IFNγR+ cells, enhanced their sensitivity to IFNγR-dependent deletion, and impaired tumor growth.

Contributed by Paula Hochman

Bystander IFN-γ activity promotes widespread and sustained cytokine signaling altering the tumor microenvironment

Using intravital microscopy and mosaic (antigen [Ag]+/Ag-) tumor models, Thibaut et al. investigated spatiotemporal effects of IFNγ in the TME. Following local antigen stimulation, IFNγ diffused and signaled extensively within the TME, affecting both tumor cells and TILs. In mosaic tumors, strong T cell-derived bystander IFNγ activity occurred in distant Ag- tumor cells, was dependent on the responding T cell concentration, and resulted in sustained STAT1 signaling. Single-cell RNAseq of human melanoma samples revealed an IFNγ signature in monocytes/macrophages, which significantly correlated with the number of IFNγ+CD8+ T cells.

Contributed by Katherine Turner

CLINICAL TRIAL: Intratumoral delivery of tavokinogene telseplasmid yields systemic immune responses in metastatic melanoma patients

In a phase II clinical trial, patients with stage III/IV melanoma were treated with intratumoral injections of tavokinogene telseplasmid (Tavo) – a plasmid encoding IL-12 – followed by electroporation. Tavo was well tolerated. Best ORR in 28 evaluable patients was 36% (18% CR) and median OS was not reached at a median follow-up of 30 months. Best ORR in treated and untreated lesions were 44% and 25%, respectively. Tavo increased TILs, T cell and NK cell activation, antigen presentation, and T cell trafficking, leading to systemic response. Tavo also increased PD-L1 expression, and 6 of 8 patients progressing on Tavo responded to subsequent pembrolizumab.

Contributed by Anna Scherer

Valproic acid attenuates CCR2-dependent tumor infiltration of monocytic myeloid-derived suppressor cells, limiting tumor progression

Xie et al. demonstrate that the antitumor effect of valproic acid (VPA), a histone deacetylase inhibitor approved as an anticonvulsant, is mediated by reduced infiltration of monocytic (M)-MDSCs and is dependent on NK and possibly T cell activity in EL4 (lymphoma) and B16-10 (melanoma) mouse models. VPA administration decreased M-MDSCs, but not PMN-MDSCs, infiltration via downregulation of CCR2 expression, and increased NK and T cells activation and infiltration. VPA enhanced the antitumor effect of anti-PD-1 therapy in both anti-PD-1-sensitive EL4 and anti-PD-1-resistant B16-10 mouse models.

Contributed by Shishir Pant

PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer

Diskin et al. studied the relatively unexplored role of PD-L1 expression on T cells and showed that PD-L1 expression on T cells is induced by JAK-STAT signaling, tumor antigen presentation, and inflammatory cues. Conditional deletion of PD-L1 in T cells in an orthotopic KPC pancreatic tumor model enhanced intratumoral T cells and immunogenic macrophage activation. Engagement of PD-L1 by PD-1 in CD4+ T cells triggered suppressive back signaling and favored TH17 over TH1-/TH2 polarization. PD-L1+ T cells also suppressed CD4+ and CD8+ T cells in the TME and induced immunosuppressive M2-like macrophage differentiation via PD-1 ligation.

Contributed by Shishir Pant

Differentiated agonistic antibody targeting CD137 eradicates large tumors without hepatotoxicity

Two agonistic antibody therapies targeting CD137 (4-1BB) resulted in hepatotoxicity and poor efficacy in the clinic. To improve these profiles, Eskiocak et al. developed CTX-471, an IgG4-Fc anti-CD137 antibody which regressed exceptionally large tumors in mouse models without inducing spleen or liver toxicities. CTX-471 binding was not competitive with, and synergized with CD137 ligand binding. Efficacy depended on both T and NK cells, as well as Fc receptor engagement with IgG4. Treatment increased colon carcinoma T cell infiltration, reduced intratumoral Tregs and exhaustion marker expression, and enriched for memory signatures.

Contributed by Alex Najibi

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.